Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Natl Cancer Inst ; 111(6): 538-549, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30859213

RESUMEN

Antibody-drug conjugates (ADCs) are complex engineered therapeutics consisting of monoclonal antibodies, directed toward tumor-associated antigens, to which highly potent cytotoxic agents are attached using chemical linkers. This targeted drug delivery strategy couples the precision of the antibody targeting moiety with the cytocidal activity of the payload, which is generally too toxic on its own to be systemically administered. In this manner, ADCs confer a means to reduce off-target toxicities in patients by limiting the exposure of normal tissues to the payload, thus broadening the potential therapeutic window compared with traditional chemotherapy. The pace of ADC development is accelerating, with the number of investigational agents in human trials having more than tripled over the past 5 years, underscoring the enthusiasm for this transformative approach to cancer treatment. Here, we review the key structural elements of ADC design (antibody, linker, and payload), highlighting critical aspects and technological advances that have affected the clinical effectiveness of this class of biopharmaceuticals. The ADC field continues to evolve, including ongoing efforts aimed at improving target selection, developing payloads with varied mechanisms of action and increased potency, designing innovative bioconjugation strategies, as well as maximizing efficacy and tolerability in patients. An overview of the current clinical trial landscape is provided, with emphasis on the clinical experience of the four ADCs to have received regulatory approval to date, as well as additional promising candidates currently in late-stage clinical development in both solid tumor and hematological malignancies.


Asunto(s)
Inmunoconjugados/administración & dosificación , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Neoplasias/metabolismo
2.
Mol Cancer Ther ; 14(11): 2422-32, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26271675

RESUMEN

The clinical benefits of chemotherapy are commonly offset by insufficient drug exposures, narrow safety margins, and/or systemic toxicities. Over recent decades, a number of conjugate-based targeting approaches designed to overcome these limitations have been explored. Here, we report on an innovative strategy that utilizes HSP90 inhibitor-drug conjugates (HDC) for directed tumor targeting of chemotherapeutic agents. STA-12-8666 is an HDC that comprises an HSP90 inhibitor fused to SN-38, the active metabolite of irinotecan. Mechanistic analyses in vitro established that high-affinity HSP90 binding conferred by the inhibitor backbone could be exploited for conjugate accumulation within tumor cells. In vivo modeling showed that the HSP90 inhibitor moiety was required for selective retention of STA-12-8666, and this enrichment promoted extended release of active SN-38 within the tumor compartment. Indeed, controlled intratumoral payload release by STA-12-8666 contributed to a broad therapeutic window, sustained biomarker activity, and remarkable degree of efficacy and durability of response in multiple cell line and patient-derived xenograft models. Overall, STA-12-8666 has been developed as a unique HDC agent that employs a distinct mechanism of targeted drug delivery to achieve potent and sustained antitumor effects. These findings identify STA-12-8666 as a promising new candidate for evaluation as novel anticancer therapeutic.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Resorcinoles/farmacología , Triazoles/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Western Blotting , Camptotecina/química , Camptotecina/farmacocinética , Camptotecina/farmacología , Línea Celular Tumoral , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Irinotecán , Ratones Endogámicos ICR , Ratones SCID , Microscopía Fluorescente , Terapia Molecular Dirigida/métodos , Neoplasias/metabolismo , Neoplasias/patología , Resorcinoles/química , Resorcinoles/farmacocinética , Inhibidores de Topoisomerasa I/administración & dosificación , Inhibidores de Topoisomerasa I/farmacocinética , Inhibidores de Topoisomerasa I/farmacología , Resultado del Tratamiento , Triazoles/administración & dosificación , Triazoles/farmacocinética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
PLoS One ; 10(5): e0127361, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25974040

RESUMEN

Systemic lupus erythematosus (SLE) is a complex, systemic autoimmune disease with a diverse range of immunological and clinical manifestations. The introduction of broad spectrum immunosuppressive therapies and better management of acute disease exacerbations have improved outcomes for lupus patients over recent years. However, these regimens are burdened by substantial toxicities and confer significantly higher risks of infection, thus there remains a significant and unmet medical need for alternative treatment options, particularly those with improved safety profiles. Heat shock protein 90 (HSP90) is a ubiquitously expressed molecular chaperone that acts as an important modulator of multiple innate and adaptive inflammatory processes. Of note, accumulating clinical and experimental evidence has implicated a role for HSP90 in the pathogenesis of SLE. Here we evaluated the potential of HSP90 as a therapeutic target for this disease using the selective small molecule inhibitor ganetespib in the well-characterized MRL/lpr autoimmune mouse model. In both the prophylactic and therapeutic dosing settings, ganetespib treatment promoted dramatic symptomatic improvements in multiple disease parameters, including suppression of autoantibody production and the preservation of renal tissue integrity and function. In addition, ganetespib exerted profound inhibitory effects on disease-related lymphadenopathy and splenomegaly, and reduced pathogenic T and B cell lineage populations in the spleen. Ganetespib monotherapy was found to be equally efficacious and tolerable when compared to an effective weekly dosing regimen of the standard-of-care immunosuppressive agent cyclophosphamide. Importantly, co-treatment of ganetespib with a sub-optimal, intermittent dosing schedule of cyclophosphamide resulted in superior therapeutic indices and maximal disease control. These findings highlight the potential of HSP90 inhibition as an alternative, and potentially complementary, strategy for therapeutic intervention in SLE. Such approaches may have important implications for disease management, particularly for limiting or preventing treatment-related toxicities, a major confounding factor in current SLE therapy.


Asunto(s)
Ciclofosfamida/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Triazoles/uso terapéutico , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Ciclofosfamida/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunosupresores/farmacología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Ratones , Bazo/efectos de los fármacos , Bazo/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Triazoles/farmacología
4.
Target Oncol ; 10(2): 235-45, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25077897

RESUMEN

Small molecule inhibitors of epidermal growth factor receptor (EGFR) tyrosine kinase activity, such as erlotinib and gefitinib, revolutionized therapy for non-small cell lung cancer (NSCLC) patients whose tumors harbor activating EGFR mutations. However, mechanisms to overcome the invariable development of acquired resistance to such agents, as well as realizing their full clinical potential within the context of wild-type EGFR (WT-EGFR) disease, remain to be established. Here, the antitumor efficacy of targeted EGFR tyrosine kinase inhibitors (TKIs) and the HSP90 inhibitor ganetespib, alone and in combination, were evaluated in NSCLC. Ganetespib potentiated the efficacy of erlotinib in TKI-sensitive, mutant EGFR-driven NCI-HCC827 xenograft tumors, with combination treatment causing significant tumor regressions. In erlotinib-resistant NCI-H1975 xenografts, concurrent administration of ganetespib overcame erlotinib resistance to significantly improve tumor growth inhibition. Ganetespib co-treatment also significantly enhanced antitumor responses to afatinib in the same model. In WT-EGFR cell lines, ganetespib potently reduced cell viability. In NCI-H1666 cells, ganetespib-induced loss of client protein expression, perturbation of oncogenic signaling pathways, and induction of apoptosis translated to robust single-agent activity in vivo. Dual ganetespib/erlotinib therapy induced regressions in NCI-H322 xenograft tumors, indicating that the sensitizing properties of ganetespib for erlotinib were conserved within the WT-EGFR setting. Mechanistically, combined ganetespib/erlotinib exposure stabilized EGFR protein levels in an inactive state and completely abrogated extracellular-signal-regulated kinase (ERK) and AKT signaling activity. Thus, selective HSP90 blockade by ganetespib represents a potentially important complementary strategy to targeted TKI inhibition alone for inducing substantial antitumor responses and overcoming resistance, in both the mutant and WT-EGFR settings.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Triazoles/farmacología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones SCID , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Cancer Res ; 12(7): 1042-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24784839

RESUMEN

UNLABELLED: Activating mutations and/or overexpression of FGFR3 are common in bladder cancer, making FGFR3 an attractive therapeutic target in this disease. In addition, FGFR3 gene rearrangements have recently been described that define a unique subset of bladder tumors. Here, a selective HSP90 inhibitor, ganetespib, induced loss of FGFR3-TACC3 fusion protein expression and depletion of multiple oncogenic signaling proteins in RT112 bladder cells, resulting in potent cytotoxicity comparable with the pan-FGFR tyrosine kinase inhibitor BGJ398. However, in contrast to BGJ398, ganetespib exerted pleiotropic effects on additional mitogenic and survival pathways and could overcome the FGFR inhibitor-resistant phenotype of FGFR3 mutant-expressing 97-7 and MHG-U3 cells. Combinatorial benefit was observed when ganetespib was used with BGJ398 both in vitro and in vivo. Interestingly, two additional FGFR3 fusion-positive lines (RT4 and SW480) retained sensitivity to HSP90 inhibitor treatment by the ansamycins 17-AAG and 17-DMAG yet displayed intrinsic resistance to ganetespib or AUY922, both second-generation resorcinol-based compounds. Both cell lines, compared with RT112, expressed considerably higher levels of endogenous UGT1A enzyme; this phenotype resulted in a rapid glucuronidation-dependent metabolism and subsequent efflux of ganetespib from SW780 cells, thus providing a mechanism to account for the lack of bioactivity. IMPLICATIONS: Pharmacologic blockade of the molecular chaperone HSP90 represents a promising approach for treating bladder tumors driven by oncogenic gene rearrangements of FGFR3. Furthermore, UDP-glucuronosyltransferase enzyme expression may serve as a predictive factor for clinical response to resorcinol-based HSP90 inhibitors.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Triazoles/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Línea Celular Tumoral , Femenino , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Distribución Aleatoria , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Triazoles/farmacocinética , Neoplasias de la Vejiga Urinaria/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Invest New Drugs ; 32(4): 577-86, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24682747

RESUMEN

The integration of targeted agents to standard cytotoxic regimens has improved outcomes for patients with colorectal cancer (CRC) over recent years; however this malignancy remains the second leading cause of cancer mortality in industrialized countries. Small molecule inhibitors of heat shock protein 90 (HSP90) are one of the most actively pursued classes of compounds for the development of new cancer therapies. Here we evaluated the activity of ganetespib, a second-generation HSP90 inhibitor, in models of CRC. Ganetespib reduced cell viability in a panel of CRC cell lines in vitro with low nanomolar potency. Mechanistically, drug treatment exerted concomitant effects on multiple oncogenic signaling pathways, cell cycle regulation, and DNA damage repair capacity to promote apoptosis. Combinations of ganetespib and low-dose ionizing radiation enhanced the radiosensitivity of HCT 116 cells and resulted in superior cytotoxic activity over either treatment alone. In vivo, the single-agent activity of ganetespib was relatively modest, suppressing HCT 116 xenograft tumor growth by approximately half. However, ganetespib significantly potentiated the antitumor efficacy of the 5-Fluorouracil (5-FU) prodrug capecitabine in HCT 116 xenografts, causing tumor regressions in a model that is intrinsically resistant to fluoropyrimidine therapy. This demonstration of combinatorial benefit afforded by an HSP90 inhibitor to a standard CRC adjuvant regimen provides an attractive new framework for the potential application of ganetespib as an investigational agent in this disease.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Fármacos Sensibilizantes a Radiaciones/farmacología , Triazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Capecitabina , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/radioterapia , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacología , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Radiación Ionizante , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Res ; 12(5): 703-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24554781

RESUMEN

UNLABELLED: Because of their pleiotropic effects on critical oncoproteins, inhibitors of HSP90 represent a promising new class of therapeutic agents for the treatment of human cancer. However, pharmacologic inactivation of HSP90 subsequently triggers a heat shock response that may mitigate the full therapeutic benefit of these compounds. To overcome this limitation, a clinically feasible method was sought to block HSP synthesis induced by the potent HSP90 inhibitor ganetespib. An immunoassay screen of 322 late-stage or clinically approved drugs was performed to uncover compounds that could block upregulation of the stress-inducible HSP70 that results as a consequence of HSP90 blockade. Interestingly, inhibitors of the phosphoinositide 3-kinase (PI3K)/mTOR class counteracted ganetespib-induced HSP70 upregulation at both the gene and protein level by suppressing nuclear translocation of heat shock factor 1 (HSF1), the dominant transcription factor controlling cellular stress responses. This effect was conserved across multiple tumor types and was found to be regulated, in part, by mTOR-dependent translational activity. Pretreatment with cycloheximide, PI3K/mTOR inhibitor, or an inhibitor of eIF4E (a translation initiation factor and downstream effector of mTOR) all reduced ganetespib-mediated nuclear HSF1 accumulation, indicating that mTOR blockade confers a negative regulatory effect on HSF1 activity. Moreover, combined therapy regimens with mTOR or dual PI3K/mTOR inhibitors potentiated the antitumor efficacy of ganetespib in multiple in vivo models. IMPLICATIONS: Collectively these data identify a novel strategy to optimize the therapeutic potential of HSP90 inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones SCID , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Triazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Res ; 74(5): 1294-300, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24556722

RESUMEN

As with many physiologic processes that become subverted during tumorigenesis, the chaperoning activity of heat shock protein 90 (HSP90) is often exploited by cancer cells to confer aberrant proliferative, survival, and/or metastatic potential. Functional inhibition of HSP90 results in the degradation of its client proteins, in turn providing a means to concomitantly disrupt multiple oncogenic signaling cascades through one molecular target. Pharmacologic blockade of HSP90 has, therefore, emerged as an innovative and multifaceted approach for the development of new antineoplastic agents. However, no HSP90 inhibitors are currently approved for cancer therapy and the full promise of this class of agents is yet to be realized. This review focuses on the preclinical activity profile of ganetespib, a potent small-molecule inhibitor of HSP90, the characterization of which has provided important frameworks for the optimal design and application of HSP90 inhibitor-based strategies in a variety of cancer types. Beyond client protein-driven tumors, ganetespib can also potentiate the effects of other molecularly targeted and standard-of-care therapeutics while simultaneously overcoming drug resistance in multiple tumor types, thereby positioning this compound as the leading HSP90 inhibitor currently under clinical development.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Triazoles/farmacología , Triazoles/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Neoplasias/metabolismo
9.
Mol Cancer Ther ; 13(2): 353-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24398428

RESUMEN

Activating BRAF kinase mutations serve as oncogenic drivers in over half of all melanomas, a feature that has been exploited in the development of new molecularly targeted approaches to treat this disease. Selective BRAF(V600E) inhibitors, such as vemurafenib, typically induce initial, profound tumor regressions within this group of patients; however, durable responses have been hampered by the emergence of drug resistance. Here, we examined the activity of ganetespib, a small-molecule inhibitor of Hsp90, in melanoma lines harboring the BRAF(V600E) mutation. Ganetespib exposure resulted in the loss of mutant BRAF expression and depletion of mitogen-activated protein kinase and AKT signaling, resulting in greater in vitro potency and antitumor efficacy compared with targeted BRAF and MAP-ERK kinase (MEK) inhibitors. Dual targeting of Hsp90 and BRAF(V600E) provided combinatorial benefit in vemurafenib-sensitive melanoma cells in vitro and in vivo. Importantly, ganetespib overcame mechanisms of intrinsic and acquired resistance to vemurafenib, the latter of which was characterized by reactivation of extracellular signal-regulated kinase (ERK) signaling. Continued suppression of BRAF(V600E) by vemurafenib potentiated sensitivity to MEK inhibitors after acquired resistance had been established. Ganetespib treatment reduced, but not abolished, elevations in steady-state ERK activity. Profiling studies revealed that the addition of a MEK inhibitor could completely abrogate ERK reactivation in the resistant phenotype, with ganetespib displaying superior combinatorial activity over vemurafenib. Moreover, ganetespib plus the MEK inhibitor TAK-733 induced tumor regressions in vemurafenib-resistant xenografts. Overall these data highlight the potential of ganetespib as a single-agent or combination treatment in BRAF(V600E)-driven melanoma, particularly as a strategy to overcome acquired resistance to selective BRAF inhibitors.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Indoles/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Triazoles/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Resistencia a Antineoplásicos/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Ratones Desnudos , Ratones SCID , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Piridonas/administración & dosificación , Piridonas/farmacología , Pirimidinonas/administración & dosificación , Pirimidinonas/farmacología , Triazoles/administración & dosificación , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Clin Cancer Res ; 20(2): 413-24, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24173541

RESUMEN

PURPOSE: Treatment options for patients with triple-negative breast cancer (TNBC) are largely limited to systemic chemotherapies, which have shown disappointing efficacy in the metastatic setting. Here, we undertook a comprehensive evaluation of the activity of ganetespib, a potent inhibitor of HSP90, in this malignancy. EXPERIMENTAL DESIGN: The antitumor and antimetastatic activity of ganetespib was investigated using TNBC cell lines and xenograft models. Combinatorial drug analyses were performed with chemotherapeutic agents and concomitant effects on DNA damage and cell-cycle disruption were assessed in vitro; antitumor efficacy was assessed in vivo. Metabolic and objective tumor responses were evaluated in patients with metastatic TNBC undergoing ganetespib treatment. RESULTS: Ganetespib simultaneously deactivated multiple oncogenic pathways to potently reduce cell viability in TNBC cell lines, and suppressed lung metastases in experimental models. Ganetespib potentiated the cytotoxic activity of doxorubicin via enhanced DNA damage and mitotic arrest, conferring superior efficacy to the doxorubicin-cyclophosphamide regimen in TNBC xenografts. Ganetespib also promoted mitotic catastrophe and apoptosis in combination with taxanes in vitro, and these effects translated to significantly improved combinatorial activity in vivo. Marked tumor shrinkage of metastatic lung and lymphatic lesions were seen in patients on ganetespib monotherapy. CONCLUSION: The preclinical activity profile and clinical evidence of tumor regression suggest that ganetespib offers considerable promise as a new therapeutic candidate to target TNBC.


Asunto(s)
Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Triazoles/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Mitosis/efectos de los fármacos , Metástasis de la Neoplasia , Estadificación de Neoplasias , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/diagnóstico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Toxicol Appl Pharmacol ; 273(2): 401-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24090817

RESUMEN

In human trials certain heat shock protein 90 (Hsp90) inhibitors, including 17-DMAG and NVP-AUY922, have caused visual disorders indicative of retinal dysfunction; others such as 17-AAG and ganetespib have not. To understand these safety profile differences we evaluated histopathological changes and exposure profiles of four Hsp90 inhibitors, with or without clinical reports of adverse ocular effects, using a rat retinal model. Retinal morphology, Hsp70 expression (a surrogate marker of Hsp90 inhibition), apoptotic induction and pharmacokinetic drug exposure analysis were examined in rats treated with the ansamycins 17-DMAG and 17-AAG, or with the second-generation compounds NVP-AUY922 and ganetespib. Both 17-DMAG and NVP-AUY922 induced strong yet restricted retinal Hsp70 up-regulation and promoted marked photoreceptor cell death 24h after the final dose. In contrast, neither 17-AAG nor ganetespib elicited photoreceptor injury. When the relationship between drug distribution and photoreceptor degeneration was examined, 17-DMAG and NVP-AUY922 showed substantial retinal accumulation, with high retina/plasma (R/P) ratios and slow elimination rates, such that 51% of 17-DMAG and 65% of NVP-AUY922 present at 30 min post-injection were retained in the retina 6h post-dose. For 17-AAG and ganetespib, retinal elimination was rapid (90% and 70% of drugs eliminated from the retina at 6h, respectively) which correlated with lower R/P ratios. These findings indicate that prolonged inhibition of Hsp90 activity in the eye results in photoreceptor cell death. Moreover, the results suggest that the retina/plasma exposure ratio and retinal elimination rate profiles of Hsp90 inhibitors, irrespective of their chemical class, may predict for ocular toxicity potential.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/metabolismo , Animales , Benzoquinonas/toxicidad , Relación Dosis-Respuesta a Droga , Proteínas HSP90 de Choque Térmico/metabolismo , Lactamas Macrocíclicas/toxicidad , Masculino , Células Fotorreceptoras/efectos de los fármacos , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Valor Predictivo de las Pruebas , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Degeneración Retiniana/patología
12.
Cancer Discov ; 3(4): 430-43, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23533265

RESUMEN

UNLABELLED: EML4-ALK gene rearrangements define a unique subset of patients with non-small cell lung carcinoma (NSCLC), and the clinical success of the anaplastic lymphoma kinase (ALK) inhibitor crizotinib in this population has become a paradigm for molecularly targeted therapy. Here, we show that the Hsp90 inhibitor ganetespib induced loss of EML4-ALK expression and depletion of multiple oncogenic signaling proteins in ALK-driven NSCLC cells, leading to greater in vitro potency, superior antitumor efficacy, and prolonged animal survival compared with results obtained with crizotinib. In addition, combinatorial benefit was seen when ganetespib was used with other targeted ALK agents both in vitro and in vivo. Importantly, ganetespib overcame multiple forms of crizotinib resistance, including secondary ALK mutations, consistent with activity seen in a patient with crizotinib-resistant NSCLC. Cancer cells driven by ALK amplification and oncogenic rearrangements of ROS1 and RET kinase genes were also sensitive to ganetespib exposure. Taken together, these results highlight the therapeutic potential of ganetespib for ALK-driven NSCLC. SIGNIFICANCE: In addition to direct kinase inhibition, pharmacologic blockade of the molecular chaperone Hsp90 is emerging as a promising approach for treating tumors driven by oncogenic rearrangements of ALK. The bioactivity profi le of ganetespib presented here underscores a new therapeutic opportunity to target ALK and overcome multiple mechanisms of resistance in patients with ALK-positive NSCLC.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/genética , Triazoles/administración & dosificación , Adulto , Quinasa de Linfoma Anaplásico , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Crizotinib , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Pirazoles/administración & dosificación , Piridinas/administración & dosificación , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
13.
PLoS One ; 6(4): e18552, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21533169

RESUMEN

There is accumulating evidence that dysregulated JAK signaling occurs in a wide variety of cancer types. In particular, mutations in JAK2 can result in the constitutive activation of STAT transcription factors and lead to oncogenic growth. JAK kinases are established Hsp90 client proteins and here we show that the novel small molecule Hsp90 inhibitor ganetespib (formerly STA-9090) exhibits potent in vitro and in vivo activity in a range of solid and hematological tumor cells that are dependent on JAK2 activity for growth and survival. Of note, ganetespib treatment results in sustained depletion of JAK2, including the constitutively active JAK2(V617F) mutant, with subsequent loss of STAT activity and reduced STAT-target gene expression. In contrast, treatment with the pan-JAK inhibitor P6 results in only transient effects on these processes. Further differentiating these modes of intervention, RNA and protein expression studies show that ganetespib additionally modulates cell cycle regulatory proteins, while P6 does not. The concomitant impact of ganetespib on both cell growth and cell division signaling translates to potent antitumor efficacy in mouse models of xenografts and disseminated JAK/STAT-driven leukemia. Overall, our findings support Hsp90 inhibition as a novel therapeutic approach for combating diseases dependent on JAK/STAT signaling, with the multimodal action of ganetespib demonstrating advantages over JAK-specific inhibitors.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Neoplasias Experimentales/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Triazoles/farmacología , Animales , División Celular/efectos de los fármacos , Genes cdc , Ratones , Neoplasias Experimentales/patología , Células Tumorales Cultivadas
14.
Cells Tissues Organs ; 185(1-3): 29-39, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17587805

RESUMEN

In addition to allowing epithelial cells to escape the structural constraints imposed by tissue architecture and adopt a phenotype more amenable to cell movement, it is now recognized that the epithelial-mesenchymal transition (EMT) may also represent a critical component permitting the progression of carcinomas towards invasive and metastatic disease. However, data supporting the actual occurrence of EMT in specific solid tumors and its relevance to the process of progression of these cancers has been scant. Despite an extensive knowledge of the genetic basis for colorectal cancer, the translation of this information into effective treatments has been limited. Clearly, there is a desperate need for new and improved therapies and since the switch to a metastatic phenotype is critical for outcome, it is of paramount importance to elucidate the biology that underlies the progression of this disease. Thus, the unique LIM 1863 model for studying the EMT of colorectal carcinoma has been used to both substantiate the importance of the transition for this cancer type and to identify molecular events that contribute to disease progression. Importantly, it has emerged that not only does EMT enhance migratory capacity, but also elicits additional selective advantages to colonic tumor cells. Specifically, the acquisition of autocrine growth factor signaling loops, mechanisms to evade apoptosis, and expression of specific integrins allowing invasive cells to interact with interstitial matrices and sustain activation of TGF-beta combine to provide a compelling new biochemical framework for understanding how EMT contributes to tumor evolution.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Epiteliales/patología , Mesodermo/patología , Línea Celular Tumoral , Citocinas/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/patología , Humanos , Integrinas/metabolismo , Mesodermo/metabolismo , Modelos Biológicos
15.
Biochem Biophys Res Commun ; 351(1): 51-6, 2006 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17052693

RESUMEN

CD36 is a transmembrane glycoprotein receptor that engages in signal transduction implicated in important physiological and pathophysiological events. CD36 in platelets has been shown physically and functionally to associate with members of the Src family of protein tyrosine kinases, Fyn, Lyn, and Yes, but the nature of this important association has never been rigorously examined. Here, we show that CD36 does not associate with Lyn through a protein-mediated interaction. In COS cells transfected with both CD36 and Lyn these molecules did not co-precipitate, suggesting a requirement for an intermediary molecule absent from the COS cells. Yeast two-hybrid analysis confirmed that the carboxylterminal cytoplasmic tail of CD36 did not bind Lyn directly, and no Lyn binding protein bound to CD36 in a cDNA library screen. Conversely, when the CD36-Lyn association seen in platelets was analysed by biophysical parameters, dissociation occurred at 37 degrees C and also by solubilisation in octylglucoside, indicative of a lipid-mediated association. Since both CD36 and Lyn are enriched in Triton X-100-insoluble rafts at the plasma membrane, these findings point to the importance of raft-associated lipids in CD36-mediated signal transduction.


Asunto(s)
Antígenos CD36/metabolismo , Metabolismo de los Lípidos/fisiología , Microdominios de Membrana/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo , Unión Proteica
16.
Cancer Res ; 65(23): 10938-45, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16322241

RESUMEN

We examined the expression and localization of p120 catenin (p120ctn) as a consequence of the epithelial to mesenchymal transition (EMT) of highly differentiated colon carcinoma cells (LIM1863 cells). This unique line grows in suspension as spheroids and undergoes an EMT within 24 hours following stimulation with transforming growth factor-beta and tumor necrosis factor-alpha. Although p120ctn expression remains stable during the EMT, its localization shifts from cell-cell junctions to the cytoplasm. Interestingly, a marked decrease in RhoA activation coincident with E-cadherin loss occurs during the EMT and correlates with the formation of a p120ctn/RhoA complex. Use of RNA interference showed that p120ctn reduction results in increased RhoA activity and a significant decrease in the motility of post-EMT cells. To determine the relevance of these findings to colorectal cancer progression, we assessed p120ctn expression by immunohistochemistry in 557 primary tumors. Of note, we observed that 53% of tumors presented cytoplasmic staining for p120ctn, and statistical analysis revealed that this localization is predictive of poor patient outcome. Cytoplasmic p120ctn correlated with later-stage tumors, significantly reduced 5- and 10-year survival times and a greater propensity for metastasis to lymph nodes compared with junctional p120ctn. We also confirmed that altered localization of p120ctn corresponded with loss or cytoplasmic localization of E-cadherin. These alterations in E-cadherin are also associated with a significant reduction in patient survival time and an increase in tumor stage and lymph node metastasis. These data provide a compelling argument for the importance of both p120ctn and the EMT itself in the progression of colorectal carcinoma.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Fosfoproteínas/metabolismo , Cadherinas/biosíntesis , Cadherinas/metabolismo , Cateninas , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Citoplasma/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Inmunohistoquímica , Uniones Intercelulares/metabolismo , Mesodermo/metabolismo , Mesodermo/patología , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Pronóstico , Interferencia de ARN , Proteína de Unión al GTP rhoA/metabolismo , Catenina delta
17.
Cell Cycle ; 4(10): 1350-2, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16123591

RESUMEN

Carcinomas, those tumors that arise from epithelial tissues, represent the most prevalent form of human malignancies. Moreover, it is the metastatic spread of these cancers, rather than the development of primary neoplasms that predominantly accounts for patient mortality. Thus, understanding the mechanisms that contribute to the process of cancer progression is of fundamental importance lo designing better therapeutic strategies for treating this disease. The epithelial-mesenchymal transition (EMT), which facilitates the process by which epithelial tumors become invasive and progress towards malignant disease, provides a superb paradigm for studying such mechanisms. Using a colon carcinoma model of EMT, it was discovered that increased expression of the integrin alphavbeta6 is elicited as a consequence of the transition. Importantly, the consequences of the elevated alphavbeta6 expression are directly linked to both tumor cell function and to the mechanism of the EMT itself. Most significantly increased alphavbeta6 expression in human tumors is a prognostic variable and one that is predictive of outcome for early-stage disease. These findings reinforce the importance of the EMT event in late stage tumorigenesis, and define alphavbeta6 as a novel therapeutic candidate for aggressive colorectal cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Integrinas/metabolismo , Mesodermo/patología , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Humanos , Mesodermo/metabolismo , Invasividad Neoplásica
18.
Cancer Biol Ther ; 4(4): 365-70, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15846061

RESUMEN

During embryonic development, epithelial cells must escape the structural constraints imposed by tissue architecture and adopt a phenotype more amenable to cell movement, a process known as the epithelial-mesenchymal transition (EMT). The progression of carcinomas to invasive and metastatic disease may also involve localized occurrences of EMT. However, data that support the actual occurrence of EMT in specific carcinomas and the relevance of this process to the progression of these tumors had been scant. This review highlights recent studies that substantiate the importance of the EMT to colorectal carcinoma. Specifically, a novel model for studying the EMT of colorectal carcinoma has been used to gain insight into the nature of the EMT itself and to identify molecular events that contribute to disease progression. Although loss of E-cadherin function is a primal event for the EMT, the expression of specific integrins such as alpha(v)beta6 as a consequence of the EMT enables invasive cells to interact with interstitial matrices and to sustain activation of TGF-beta. Of note, alpha(v)beta6 expression in tumors is a marker of cells that have undergone an EMT and it is prognostic for tumors that will progress more rapidly to terminal disease. The EMT also induces autocrine signaling involving VEGF and Flt-1 that enable invasive cells to become 'self-sufficient' for survival. Thus, the EMT appears to be an integral component of colorectal cancer progression and its analysis can yield novel targets for prognosis and therapy.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Animales , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor , Progresión de la Enfermedad , Epitelio/metabolismo , Epitelio/patología , Humanos , Integrinas/metabolismo , Mesodermo/metabolismo , Mesodermo/patología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Clin Invest ; 115(2): 339-47, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15668738

RESUMEN

We used a spheroid model of colon carcinoma to analyze integrin dynamics as a function of the epithelial-mesenchymal transition (EMT), a process that provides a paradigm for understanding how carcinoma cells acquire a more aggressive phenotype. This EMT involves transcriptional activation of the beta6 integrin subunit and a consequent induction of alphavbeta6 expression. This integrin enhances the tumorigenic properties of colon carcinoma, including activation of autocrine TGF-beta and migration on interstitial fibronectin. Importantly, this study validates the clinical relevance of the EMT. Kaplan-Meier analysis of beta6 expression in 488 colorectal carcinomas revealed a striking reduction in median survival time of patients with high beta6 expression. Elevated receptor expression did not simply reflect increasing tumor stage, since log-rank analysis showed a more significant impact on the survival of patients with early-stage, as opposed to late-stage, disease. Cox regression analysis confirmed that this integrin is an independent variable for these tumors. These findings define the alphavbeta6 integrin as an important risk factor for early-stage disease and a novel therapeutic candidate for colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma/metabolismo , Carcinoma/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Integrina alfa5/biosíntesis , Cadenas beta de Integrinas/biosíntesis , Animales , Comunicación Autocrina , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/patología , Mesodermo/patología , Ratones , Ratones Desnudos , Invasividad Neoplásica , Pronóstico , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
20.
Future Oncol ; 1(6): 821-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16556062

RESUMEN

Recent advances in the treatment of colorectal cancer have resulted from a shift towards the use of molecular targeting strategies, with the identification of specific antigens critical for the development and progression of these tumors allowing the rational design of new therapeutic agents. The integrin receptor family provides very attractive targets for this 'magic bullet' approach, as they play key roles at all points during the malignant evolution of tumors, influencing growth, progression, angiogenesis and metastasis. One integrin in particular, alphaVbeta6, was recently shown to be upregulated in a model of colorectal tumor progression. Importantly, the consequences of this elevated receptor expression not only impact tumor cell function, but also indicate that alphaVbeta6 is a prognostic variable for colorectal cancer and one that is predictive of patient outcome in early-stage disease. Taken together with its normal expression pattern and known biologic functions, alphaVbeta6 thus emerges as a novel therapeutic candidate.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias Colorrectales/metabolismo , Células Epiteliales/patología , Matriz Extracelular/patología , Integrinas/metabolismo , Mesodermo/patología , Neoplasias Colorrectales/patología , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Humanos , Mesodermo/metabolismo , Invasividad Neoplásica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA